1
|
Todorova E, Kavrakova A, Derimachkovski G, Georgieva B, Odzhakov F, Bachurska S, Terziev I, Boyadzhieva ME, Valkov T, Popov E, Slavov C, Tourtourikov I, Mitev V, Todorova A. Human Herpes Virus Genotype and Immunological Gene Expression Profile in Prostate Cancer with Prominent Inflammation. Int J Mol Sci 2025; 26:4945. [PMID: 40430084 DOI: 10.3390/ijms26104945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 05/15/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
We aim to investigate the role of the Herpesviridae family (HHV) in the onset and progression of prostate cancer (PCa) and to profile the local PCa immunological status. A total of 116 "tru-cut" biopsies (58 PCa and 58 benign prostatic hyperplasia [BPH]) and 49 formalin-fixed paraffin-embedded (FFPE) instances of PCa were analysed using real-time qPCR and histological examination. Infection with CMV, EBV, HHV6, and HHV7 was detected in 11.5% of the "tru-cut" biopsies (25.9% in BPH and 6.9% in the PCa group). In the formalin-fixed paraffin-embedded (FFPE) samples, infection was detected in 69.4% of the patients, with individual rates of EBV (47%), HHV6 (38%), HHV7 (41%), CMV (2.9%), HSV2 (2.9%), and VZV (5.8%). In the HHV-infected PCa cases, the histopathological landscape included intratumor lymphocyte infiltration with fibrosis and necrosis, periductal chronic inflammatory reaction and granulomatous lesions with foci of abscesses and necrosis, as well as inflammatory infiltration, chronic lymphadenitis, prostatic intraepithelial atrophy (PIA), and high-grade prostatic intraepithelial neoplasia (HGPIN). The majority of HHV-infected PCa patients were predominantly classified as grade G3/G4/G5 tumours, exhibiting perineural, perivascular, and lymphovascular invasion, seminal vesicle invasion, senile vesicle amyloidosis, and lymph node metastasis. Statistical analysis demonstrated a significant association between HHV infection and PCa (χ2 ≈ 20.3, df = 1, p < 0.0001; Fisher's exact test, p < 0.0001) with an odds ratio of 6.50 (95% CI: 2.80-15.12). These findings suggest that long-term HHV infection could contribute to a complicated and potentially altered immune PCa tumour environment due to inflammation. This may serve as a predictor of aggressive disease progression.
Collapse
Affiliation(s)
- Elena Todorova
- Department of Medical Chemistry and Biochemistry, MU-Sofia, 1431 Sofia, Bulgaria
- Genetic and Medico-Diagnostic Laboratory "Genica", 1463 Sofia, Bulgaria
| | - Anita Kavrakova
- Department of Medical Chemistry and Biochemistry, MU-Sofia, 1431 Sofia, Bulgaria
- Genetic and Medico-Diagnostic Laboratory "Genica", 1463 Sofia, Bulgaria
| | | | - Bilyana Georgieva
- Department of Medical Chemistry and Biochemistry, MU-Sofia, 1431 Sofia, Bulgaria
| | - Feodor Odzhakov
- Department of General and Clinical Pathology, University Specialized Hospital for Oncology, 1756 Sofia, Bulgaria
| | - Svitlana Bachurska
- Department of General and Clinical Pathology, University Specialized Hospital for Oncology, 1756 Sofia, Bulgaria
| | - Ivan Terziev
- Department of General and Clinical Pathology, UMHAT "Tsaritsa Ioanna-ISUL", 1527 Sofia, Bulgaria
| | - Maria-Elena Boyadzhieva
- Department of General and Clinical Pathology, UMHAT "Tsaritsa Ioanna-ISUL", 1527 Sofia, Bulgaria
| | - Trifon Valkov
- Department of Infectious Diseases, MU-Sofia, Prof. Ivan Kirov Hospital, 1606 Sofia, Bulgaria
| | - Elenko Popov
- Department of Urology, UMHAT "Tsaritsa Ioanna-ISUL", MU-Sofia, 1527 Sofia, Bulgaria
| | - Chavdar Slavov
- Department of Urology, UMHAT "Tsaritsa Ioanna-ISUL", MU-Sofia, 1527 Sofia, Bulgaria
| | - Ivan Tourtourikov
- Department of Medical Chemistry and Biochemistry, MU-Sofia, 1431 Sofia, Bulgaria
- Genetic and Medico-Diagnostic Laboratory "Genica", 1463 Sofia, Bulgaria
| | - Vanyo Mitev
- Department of Medical Chemistry and Biochemistry, MU-Sofia, 1431 Sofia, Bulgaria
| | - Albena Todorova
- Department of Medical Chemistry and Biochemistry, MU-Sofia, 1431 Sofia, Bulgaria
- Genetic and Medico-Diagnostic Laboratory "Genica", 1463 Sofia, Bulgaria
| |
Collapse
|
2
|
Khan F, Müller TR, Kasmapour B, Ynga-Durand MA, Eiz-Vesper B, von Einem J, Busch DH, Cicin-Sain L. Dynamic monitoring of viral gene expression reveals rapid antiviral effects of CD8 T cells recognizing the HCMV-pp65 antigen. Front Immunol 2024; 15:1439184. [PMID: 39104541 PMCID: PMC11299495 DOI: 10.3389/fimmu.2024.1439184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 06/24/2024] [Indexed: 08/07/2024] Open
Abstract
Introduction Human Cytomegalovirus (HCMV) is a betaherpesvirus that causes severe disease in immunocompromised transplant recipients. Immunotherapy with CD8 T cells specific for HCMV antigens presented on HLA class-I molecules is explored as strategy for long-term relief to such patients, but the antiviral effectiveness of T cell preparations cannot be efficiently predicted by available methods. Methods We developed an Assay for Rapid Measurement of Antiviral T-cell Activity (ARMATA) by real-time automated fluorescent microscopy and used it to study the ability of CD8 T cells to neutralize HCMV and control its spread. As a proof of principle, we used TCR-transgenic T cells specific for the immunodominant HLA-A02-restricted tegumental phosphoprotein pp65. pp65 expression follows an early/late kinetic, but it is not clear at which stage of the virus cycle it acts as an antigen. We measured control of HCMV infection by T cells as early as 6 hours post infection (hpi). Results The timing of the antigen recognition indicated that it occurred before the late phase of the virus cycle, but also that virion-associated pp65 was not recognized during virus entry into cells. Monitoring of pp65 gene expression dynamics by reporter fluorescent genes revealed that pp65 was detectable as early as 6 hpi, and that a second and much larger bout of expression occurs in the late phase of the virus cycle by 48 hpi. Since transgenic (Tg)-pp65 specific CD8 T cells were activated even when DNA replication was blocked, our data argue that pp65 acts as an early virus gene for immunological purposes. Discussion ARMATA does not only allow same day identification of antiviral T-cell activity, but also provides a method to define the timing of antigen recognition in the context of HCMV infection.
Collapse
Affiliation(s)
- Fawad Khan
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- German Center for Infection Research (DZIF), Partner site Hannover/Braunschweig, Braunschweig, Germany
- Centre for Individualized Infection Medicine (CIIM), a Joint Venture of Helmholtz Centre for Infection Research and Hannover Medical School, Hannover, Germany
| | - Thomas R. Müller
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
- German Center for Infection Research (DZIF), Partner site Munich, Munich, Germany
| | - Bahram Kasmapour
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine (CIIM), a Joint Venture of Helmholtz Centre for Infection Research and Hannover Medical School, Hannover, Germany
| | - Mario Alberto Ynga-Durand
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine (CIIM), a Joint Venture of Helmholtz Centre for Infection Research and Hannover Medical School, Hannover, Germany
| | - Britta Eiz-Vesper
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Jens von Einem
- Institute of Virology, University Hospital Ulm, Ulm, Germany
| | - Dirk H. Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
- German Center for Infection Research (DZIF), Partner site Munich, Munich, Germany
| | - Luka Cicin-Sain
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- German Center for Infection Research (DZIF), Partner site Hannover/Braunschweig, Braunschweig, Germany
- Centre for Individualized Infection Medicine (CIIM), a Joint Venture of Helmholtz Centre for Infection Research and Hannover Medical School, Hannover, Germany
| |
Collapse
|
3
|
Santamorena MM, Tischer-Zimmermann S, Bonifacius A, Mireisz CNM, Costa B, Khan F, Kulkarni U, Lauruschkat CD, Sampaio KL, Stripecke R, Blasczyk R, Maecker-Kolhoff B, Kraus S, Schlosser A, Cicin-Sain L, Kalinke U, Eiz-Vesper B. Engineered HCMV-infected APCs enable the identification of new immunodominant HLA-restricted epitopes of anti-HCMV T-cell immunity. HLA 2024; 103:e15541. [PMID: 38923358 DOI: 10.1111/tan.15541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 04/24/2024] [Accepted: 05/07/2024] [Indexed: 06/28/2024]
Abstract
Complications due to HCMV infection or reactivation remain a challenging clinical problem in immunocompromised patients, mainly due to insufficient or absent T-cell functionality. Knowledge of viral targets is crucial to improve monitoring of high-risk patients and optimise antiviral T-cell therapy. To expand the epitope spectrum, genetically-engineered dendritic cells (DCs) and fibroblasts were designed to secrete soluble (s)HLA-A*11:01 and infected with an HCMV mutant lacking immune evasion molecules (US2-6 + 11). More than 700 HLA-A*11:01-restricted epitopes, including more than 50 epitopes derived from a broad range of HCMV open-reading-frames (ORFs) were identified by mass spectrometry and screened for HLA-A*11:01-binding using established prediction tools. The immunogenicity of the 24 highest scoring new candidates was evaluated in vitro in healthy HLA-A*11:01+/HCMV+ donors. Thus, four subdominant epitopes and one immunodominant epitope, derived from the anti-apoptotic protein UL36 and ORFL101C (A11SAL), were identified. Their HLA-A*11:01 complex stability was verified in vitro. In depth analyses revealed highly proliferative and cytotoxic memory T-cell responses against A11SAL, with T-cell responses comparable to the immunodominant HLA-A*02:01-restricted HCMVpp65NLV epitope. A11SAL-specific T cells were also detectable in vivo in immunosuppressed transplant patients and shown to be effective in an in vitro HCMV-infection model, suggesting their crucial role in inhibiting viral replication and improvement of patient's outcome. The developed in vitro pipeline is the first to utilise genetically-engineered DCs to identify naturally presented immunodominant HCMV-derived epitopes. It therefore offers advantages over in silico predictions, is transferable to other HLA alleles, and will significantly expand the repertoire of viral targets to improve therapeutic options.
Collapse
Affiliation(s)
- Maria Michela Santamorena
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School (MHH), Hannover, Germany
| | - Sabine Tischer-Zimmermann
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School (MHH), Hannover, Germany
| | - Agnes Bonifacius
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School (MHH), Hannover, Germany
- German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Hannover, Germany
| | - Chiara Noemi-Marie Mireisz
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Wuerzburg, Wuerzburg, Germany
| | - Bibiana Costa
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Fawad Khan
- Immune Ageing and Chronic Infection, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Upasana Kulkarni
- Immune Ageing and Chronic Infection, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | | | - Renata Stripecke
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf, Center for Molecular Medicine Cologne (CMMC), Institute of Translational Immuno-oncology, Cologne, Germany
- German Center for Infections Research (DZIF) Bonn-Cologne, Cologne, Germany
| | - Rainer Blasczyk
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School (MHH), Hannover, Germany
| | - Britta Maecker-Kolhoff
- German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Hannover, Germany
- German Center for Infections Research (DZIF) Bonn-Cologne, Cologne, Germany
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Sabrina Kraus
- Department of Internal Medicine II, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Andreas Schlosser
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Wuerzburg, Wuerzburg, Germany
| | - Luka Cicin-Sain
- German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Hannover, Germany
- Immune Ageing and Chronic Infection, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Cluster of Excellence - Resolving Infection Susceptibility (RESIST, EXC 2155), Hannover Medical School, Hannover, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
- Cluster of Excellence - Resolving Infection Susceptibility (RESIST, EXC 2155), Hannover Medical School, Hannover, Germany
| | - Britta Eiz-Vesper
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School (MHH), Hannover, Germany
- German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Hannover, Germany
| |
Collapse
|
4
|
Holtappels R, Büttner JK, Freitag K, Reddehase MJ, Lemmermann NA. Modulation of cytomegalovirus immune evasion identifies direct antigen presentation as the predominant mode of CD8 T-cell priming during immune reconstitution after hematopoietic cell transplantation. Front Immunol 2024; 15:1355153. [PMID: 38426094 PMCID: PMC10902149 DOI: 10.3389/fimmu.2024.1355153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/01/2024] [Indexed: 03/02/2024] Open
Abstract
Cytomegalovirus (CMV) infection is the most critical infectious complication in recipients of hematopoietic cell transplantation (HCT) in the period between a therapeutic hematoablative treatment and the hematopoietic reconstitution of the immune system. Clinical investigation as well as the mouse model of experimental HCT have consistently shown that timely reconstitution of antiviral CD8 T cells is critical for preventing CMV disease in HCT recipients. Reconstitution of cells of the T-cell lineage generates naïve CD8 T cells with random specificities among which CMV-specific cells need to be primed by presentation of viral antigen for antigen-specific clonal expansion and generation of protective antiviral effector CD8 T cells. For CD8 T-cell priming two pathways are discussed: "direct antigen presentation" by infected professional antigen-presenting cells (pAPCs) and "antigen cross-presentation" by uninfected pAPCs that take up antigenic material derived from infected tissue cells. Current view in CMV immunology favors the cross-priming hypothesis with the argument that viral immune evasion proteins, known to interfere with the MHC class-I pathway of direct antigen presentation by infected cells, would inhibit the CD8 T-cell response. While the mode of antigen presentation in the mouse model of CMV infection has been studied in the immunocompetent host under genetic or experimental conditions excluding either pathway of antigen presentation, we are not aware of any study addressing the medically relevant question of how newly generated naïve CD8 T cells become primed in the phase of lympho-hematopoietic reconstitution after HCT. Here we used the well-established mouse model of experimental HCT and infection with murine CMV (mCMV) and pursued the recently described approach of up- or down-modulating direct antigen presentation by using recombinant viruses lacking or overexpressing the central immune evasion protein m152 of mCMV, respectively. Our data reveal that the magnitude of the CD8 T-cell response directly reflects the level of direct antigen presentation.
Collapse
Affiliation(s)
- Rafaela Holtappels
- Institute for Virology and Research Center for Immunotherapy (FZI) at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Julia K. Büttner
- Institute for Virology and Research Center for Immunotherapy (FZI) at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Kirsten Freitag
- Institute for Virology and Research Center for Immunotherapy (FZI) at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matthias J. Reddehase
- Institute for Virology and Research Center for Immunotherapy (FZI) at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Niels A. Lemmermann
- Institute for Virology and Research Center for Immunotherapy (FZI) at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute of Virology, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
5
|
Chaudhry MZ, Borkner L, Kulkarni U, Berberich-Siebelt F, Cicin-Sain L. NFAT signaling is indispensable for persistent memory responses of MCMV-specific CD8+ T cells. PLoS Pathog 2024; 20:e1012025. [PMID: 38346075 PMCID: PMC10890734 DOI: 10.1371/journal.ppat.1012025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 02/23/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Cytomegalovirus (CMV) induces a unique T cell response, where antigen-specific populations do not contract, but rather inflate during viral latency. It has been proposed that subclinical episodes of virus reactivation feed the inflation of CMV-specific memory cells by intermittently engaging T cell receptors (TCRs), but evidence of TCR engagement has remained lacking. Nuclear factor of activated T cells (NFAT) is a family of transcription factors, where NFATc1 and NFATc2 signal downstream of TCR in mature T lymphocytes. We show selective impacts of NFATc1 and/or NFATc2 genetic ablations on the long-term inflation of MCMV-specific CD8+ T cell responses despite largely maintained responses to acute infection. NFATc1 ablation elicited robust phenotypes in isolation, but the strongest effects were observed when both NFAT genes were missing. CMV control was impaired only when both NFATs were deleted in CD8+ T cells used in adoptive immunotherapy of immunodeficient mice. Transcriptome analyses revealed that T cell intrinsic NFAT is not necessary for CD8+ T cell priming, but rather for their maturation towards effector-memory and in particular the effector cells, which dominate the pool of inflationary cells.
Collapse
Affiliation(s)
- M. Zeeshan Chaudhry
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Lisa Borkner
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Upasana Kulkarni
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Luka Cicin-Sain
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine, a joint venture of Helmholtz Centre for Infection Research and Medical School Hannover, Hannover, Germany
| |
Collapse
|
6
|
Büttner JK, Becker S, Fink A, Brinkmann MM, Holtappels R, Reddehase MJ, Lemmermann NA. Direct antigen presentation is the canonical pathway of cytomegalovirus CD8 T-cell priming regulated by balanced immune evasion ensuring a strong antiviral response. Front Immunol 2023; 14:1272166. [PMID: 38149242 PMCID: PMC10749961 DOI: 10.3389/fimmu.2023.1272166] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 11/30/2023] [Indexed: 12/28/2023] Open
Abstract
CD8 T cells are important antiviral effectors in the adaptive immune response to cytomegaloviruses (CMV). Naïve CD8 T cells can be primed by professional antigen-presenting cells (pAPCs) alternatively by "direct antigen presentation" or "antigen cross-presentation". In the case of direct antigen presentation, viral proteins are expressed in infected pAPCs and enter the classical MHC class-I (MHC-I) pathway of antigen processing and presentation of antigenic peptides. In the alternative pathway of antigen cross-presentation, viral antigenic material derived from infected cells of principally any cell type is taken up by uninfected pAPCs and eventually also fed into the MHC class-I pathway. A fundamental difference, which can be used to distinguish between these two mechanisms, is the fact that viral immune evasion proteins that interfere with the cell surface trafficking of peptide-loaded MHC-I (pMHC-I) complexes are absent in cross-presenting uninfected pAPCs. Murine cytomegalovirus (mCMV) models designed to disrupt either of the two presentation pathways revealed that both are possible in principle and can substitute each other. Overall, however, the majority of evidence has led to current opinion favoring cross-presentation as the canonical pathway. To study priming in the normal host genetically competent in both antigen presentation pathways, we took the novel approach of enhancing or inhibiting direct antigen presentation by using recombinant viruses lacking or overexpressing a key mCMV immune evasion protein. Against any prediction, the strongest CD8 T-cell response was elicited under the condition of intermediate direct antigen presentation, as it exists for wild-type virus, whereas the extremes of enhanced or inhibited direct antigen presentation resulted in an identical and weaker response. Our findings are explained by direct antigen presentation combined with a negative feedback regulation exerted by the newly primed antiviral effector CD8 T cells. This insight sheds a completely new light on the acquisition of viral immune evasion genes during virus-host co-evolution.
Collapse
Affiliation(s)
- Julia K. Büttner
- Institute for Virology and Research Center for Immunotherapy (FZI) at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Sara Becker
- Institute of Virology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Annette Fink
- Institute for Virology and Research Center for Immunotherapy (FZI) at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Melanie M. Brinkmann
- Institute of Genetics, Technische Universität Braunschweig, Braunschweig, Germany
- Virology and Innate Immunity Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Rafaela Holtappels
- Institute for Virology and Research Center for Immunotherapy (FZI) at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matthias J. Reddehase
- Institute for Virology and Research Center for Immunotherapy (FZI) at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Niels A. Lemmermann
- Institute for Virology and Research Center for Immunotherapy (FZI) at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute of Virology, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
7
|
Holtappels R, Becker S, Hamdan S, Freitag K, Podlech J, Lemmermann NA, Reddehase MJ. Immunotherapy of cytomegalovirus infection by low-dose adoptive transfer of antiviral CD8 T cells relies on substantial post-transfer expansion of central memory cells but not effector-memory cells. PLoS Pathog 2023; 19:e1011643. [PMID: 37972198 PMCID: PMC10688903 DOI: 10.1371/journal.ppat.1011643] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/30/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023] Open
Abstract
Cytomegaloviruses (CMVs) are host species-specific in their replication. It is a hallmark of all CMVs that productive primary infection is controlled by concerted innate and adaptive immune responses in the immunocompetent host. As a result, the infection usually passes without overt clinical symptoms and develops into latent infection, referred to as "latency". During latency, the virus is maintained in a non-replicative state from which it can reactivate to productive infection under conditions of waning immune surveillance. In contrast, infection of an immunocompromised host causes CMV disease with viral multiple-organ histopathology resulting in organ failure. Primary or reactivated CMV infection of hematopoietic cell transplantation (HCT) recipients in a "window of risk" between therapeutic hemato-ablative leukemia therapy and immune system reconstitution remains a clinical challenge. Studies in the mouse model of experimental HCT and infection with murine CMV (mCMV), followed by clinical trials in HCT patients with human CMV (hCMV) reactivation, have revealed a protective function of virus-specific CD8 T cells upon adoptive cell transfer (AT). Memory CD8 T cells derived from latently infected hosts are a favored source for immunotherapy by AT. Strikingly low numbers of these cells were found to prevent CMV disease, suggesting either an immediate effector function of few transferred cells or a clonal expansion generating high numbers of effector cells. In the murine model, the memory population consists of resting central memory T cells (TCM), as well as of conventional effector-memory T cells (cTEM) and inflationary effector-memory T cells (iTEM). iTEM increase in numbers over time in the latently infected host, a phenomenon known as 'memory inflation' (MI). They thus appeared to be a promising source for use in immunotherapy. However, we show here that iTEM contribute little to the control of infection after AT, which relies almost entirely on superior proliferative potential of TCM.
Collapse
Affiliation(s)
- Rafaela Holtappels
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Sara Becker
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Institute of Virology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Sara Hamdan
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Kirsten Freitag
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Jürgen Podlech
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Niels A. Lemmermann
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Institute of Virology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Matthias J. Reddehase
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
8
|
Kutle I, Dittrich A, Wirth D. Mouse Models for Human Herpesviruses. Pathogens 2023; 12:953. [PMID: 37513800 PMCID: PMC10384569 DOI: 10.3390/pathogens12070953] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/13/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
More than one hundred herpesviruses have been isolated from different species so far, with nine infecting humans. Infections with herpesviruses are characterized by life-long latency and represent a significant challenge for human health. To investigate the consequences of infections and identify novel treatment options, in vivo models are of particular relevance. The mouse has emerged as an economical small animal model to investigate herpesvirus infections. However, except for herpes simplex viruses (HSV-1, HSV-2), human herpesviruses cannot infect mice. Three natural herpesviruses have been identified in mice: mouse-derived cytomegalovirus (MCMV), mouse herpesvirus 68 (MHV-68), and mouse roseolovirus (MRV). These orthologues are broadly used to investigate herpesvirus infections within the natural host. In the last few decades, immunocompromised mouse models have been developed, allowing the functional engraftment of various human cells and tissues. These xenograft mice represent valuable model systems to investigate human-restricted viruses, making them particularly relevant for herpesvirus research. In this review, we describe the various mouse models used to study human herpesviruses, thereby highlighting their potential and limitations. Emphasis is laid on xenograft mouse models, covering the development and refinement of immune-compromised mice and their application in herpesvirus research.
Collapse
Affiliation(s)
- Ivana Kutle
- Research Group Model Systems for Infection, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Anne Dittrich
- Research Group Model Systems for Infection, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
- InSCREENeX GmbH, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Dagmar Wirth
- Research Group Model Systems for Infection, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
9
|
Abstract
Human cytomegalovirus (HCMV) is a prevalent herpesvirus, infecting the majority of the human population. Like other herpesviruses, it causes lifelong infection through the establishment of latency. Although reactivation from latency can cause significant morbidity and mortality in immunocompromised hosts, our understanding of HCMV latency and how it is maintained remains limited. Here, we discuss the characterized latency reservoir in hematopoietic cells in the bone marrow and the gaps in our knowledge of mechanisms that facilitate HCMV genome maintenance in dividing cells. We further review clinical evidence that strongly suggests the tissue origin of HCMV reactivation, and we outline similarities to murine cytomegalovirus where latency in tissue-resident cells has been demonstrated. Overall, we think these observations call for a rethinking of HCMV latency reservoirs and point to potential sources of HCMV latency that reside in tissues.
Collapse
Affiliation(s)
- Michal Schwartz
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Noam Stern-Ginossar
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
10
|
Hamdan S, Reddehase MJ, Holtappels R. Cytomegalovirus immune evasion sets the functional avidity threshold for protection by CD8 T cells. Med Microbiol Immunol 2023; 212:153-163. [PMID: 35364731 PMCID: PMC10085950 DOI: 10.1007/s00430-022-00733-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/15/2022] [Indexed: 02/07/2023]
Abstract
Conflicting hallmarks are attributed to cytomegalovirus (CMV) infections. CMVs are viewed as being master tacticians in "immune evasion" by subverting essentially all pathways of innate and adaptive immunity. On the other hand, CMV disease is undeniably restricted to the immunologically immature or immunocompromised host, whereas an intact immune system prevents virus spread, cytopathogenic tissue infection, and thus pathological organ manifestations. Therefore, the popular term "immune evasion" is apparently incongruous with the control of CMV infections in the immunocompetent human host as well as in experimental non-human primate and rodent models. Here, we review recent work from the mouse model that resolves this obvious discrepancy for the example of the virus-specific CD8 T-cell response. Immune evasion proteins encoded by murine CMV (mCMV) interfere with the cell surface trafficking of antigenic peptide-loaded MHC class-I (pMHC-I) complexes and thereby reduce their numbers available for interaction with T-cell receptors of CD8 T cells; but this inhibition is incomplete. As a consequence, while CD8 T cells with low interaction avidity fail to receive sufficient signaling for triggering their antiviral effector function in the presence of immune evasion proteins in infected cells, a few pMHC-I complexes that escape to the cell surface are sufficient for sensitizing high-avidity CD8 T cells. It is thus proposed that the function of immune evasion proteins is to raise the avidity threshold for activation, so that in the net result, only high-avidity cells can protect. An example showing that immune evasion proteins can make the difference between life and death is the lacking control of infection in a mouse model of MHC-I histoincompatible hematopoietic cell transplantation (allogeneic-HCT). In this model, only low-avidity CD8 T cells become reconstituted by HCT and almost all infected HCT recipients die of multiple-organ CMV disease when immune evasion proteins are expressed. In contrast, lowering the avidity threshold for antigen recognition by deletion of immune evasion proteins allowed control of infection and rescued from death.
Collapse
Affiliation(s)
- Sara Hamdan
- Institute for Virology and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Strasse 67, Hochhaus Am Augustusplatz, 55131, Mainz, Germany
| | - Matthias J Reddehase
- Institute for Virology and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Strasse 67, Hochhaus Am Augustusplatz, 55131, Mainz, Germany
| | - Rafaela Holtappels
- Institute for Virology and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Strasse 67, Hochhaus Am Augustusplatz, 55131, Mainz, Germany.
| |
Collapse
|
11
|
Latent CMV infection of Lymphatic endothelial cells is sufficient to drive CD8 T cell memory inflation. PLoS Pathog 2023; 19:e1010351. [PMID: 36689486 PMCID: PMC9894547 DOI: 10.1371/journal.ppat.1010351] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 02/02/2023] [Accepted: 12/21/2022] [Indexed: 01/24/2023] Open
Abstract
CMV, a ubiquitous herpesvirus, elicits an extraordinarily large T cell response that is sustained or increases over time, a phenomenon termed 'memory inflation.' Remarkably, even latent, non-productive infection can drive memory inflation. Despite intense research on this phenomenon, the infected cell type(s) involved are unknown. To identify the responsible cell type(s), we designed a Cre-lox murine CMV (MCMV) system, where a spread-deficient (ΔgL) virus expresses recombinant SIINFEKL only in Cre+ host cells. We found that latent infection of endothelial cells (ECs), but not dendritic cells (DCs) or hepatocytes, was sufficient to drive CD8 T cell memory inflation. Infection of Lyve-1-Cre and Prox1-CreERT2 mice revealed that amongst EC subsets, infection of lymphatic ECs was sufficient. Genetic ablation of β2m on lymphatic ECs did not prevent inflation, suggesting another unidentified cell type can also present antigen to CD8 T cells during latency. This novel system definitively shows that antigen presentation by lymphatic ECs drives robust CD8 T cell memory inflation.
Collapse
|
12
|
White TM, Bonavita CM, Stanfield BA, Farrell HE, Davis-Poynter NJ, Cardin RD. The CMV-encoded G protein-coupled receptors M33 and US28 play pleiotropic roles in immune evasion and alter host T cell responses. Front Immunol 2022; 13:1047299. [PMID: 36569845 PMCID: PMC9768342 DOI: 10.3389/fimmu.2022.1047299] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022] Open
Abstract
Introduction Human cytomegalovirus (HCMV) is a global health threat due to its ubiquity and lifelong persistence in infected people. During latency, host CD8+ T cell responses to HCMV continue to increase in a phenomenon known as memory inflation. We used murine CMV (MCMV) as a model for HCMV to characterize the memory inflation response to wild-type MCMV (KP) and a latency-defective mutant (ΔM33stop), which lacks M33, an MCMV chemokine receptor homolog. M33 is essential for normal reactivation from latency and this was leveraged to determine whether reactivation in vivo contributes to T cell memory inflation. Methods Mice were infected with wild-type or mutant MCMV and T cell responses were analyzed by flow cytometry at acute and latent time points. Ex vivo reactivation and cytotoxicity assays were carried out to further investigate immunity and virus replication. Quantitative reverse-transcriptase polymerase chain reaction (q-RTPCR) was used to examine gene expression during reactivation. MHC expression on infected cells was analyzed by flow cytometry. Finally, T cells were depleted from latently-infected B cell-deficient mice to examine the in vivo difference in reactivation between wild-type and ΔM33stop. Results We found that ΔM33stop triggers memory inflation specific for peptides derived from the immediate-early protein IE1 but not the early protein m164, in contrast to wild-type MCMV. During ex vivo reactivation, gene expression in DM33stop-infected lung tissues was delayed compared to wild-type virus. Normal gene expression was partially rescued by substitution of the HCMV US28 open reading frame in place of the M33 gene. In vivo depletion of T cells in immunoglobulin heavy chain-knockout mice resulted in reactivation of wild-type MCMV, but not ΔM33stop, confirming the role of M33 during reactivation from latency. Further, we found that M33 induces isotype-specific downregulation of MHC class I on the cell surface suggesting previously unappreciated roles in immune evasion. Discussion Our results indicate that M33 is more polyfunctional than previously appreciated. In addition to its role in reactivation, which had been previously described, we found that M33 alters viral gene expression, host T cell memory inflation, and MHC class I expression. US28 was able to partially complement most functions of M33, suggesting that its role in HCMV infection may be similarly pleotropic.
Collapse
Affiliation(s)
- Timothy M. White
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, LA, United States
| | - Cassandra M. Bonavita
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, LA, United States
| | - Brent A. Stanfield
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, LA, United States
| | - Helen E. Farrell
- School of Chemistry and Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | | | - Rhonda D. Cardin
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, LA, United States,*Correspondence: Rhonda D. Cardin,
| |
Collapse
|
13
|
Rozman B, Nachshon A, Levi Samia R, Lavi M, Schwartz M, Stern-Ginossar N. Temporal dynamics of HCMV gene expression in lytic and latent infections. Cell Rep 2022; 39:110653. [PMID: 35417700 PMCID: PMC9035752 DOI: 10.1016/j.celrep.2022.110653] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/19/2022] [Accepted: 03/18/2022] [Indexed: 01/10/2023] Open
Abstract
During productive human cytomegalovirus (HCMV) infection, viral genes are expressed in a coordinated cascade that conventionally relies on the dependencies of viral genes on protein synthesis and viral DNA replication. By contrast, the transcriptional landscape of HCMV latency is poorly understood. Here, we examine viral gene expression dynamics during the establishment of both productive and latent HCMV infections. We redefine HCMV gene expression kinetics during productive infection and reveal that viral gene regulation does not represent a simple sequential cascade; many viral genes are regulated by multiple independent modules. Using our improved gene expression classification combined with transcriptome-wide measurements of the effects of a wide array of epigenetic inhibitors on viral gene expression during latency, we show that a defining feature of latency is the unique repression of immediate-early (IE) genes. Altogether, we recharacterize HCMV gene expression kinetics and reveal governing principles of lytic and latent gene expression. Redefining HCMV gene expression cascade during productive infection Many viral genes are regulated by multiple independent modules Diverse inhibitors induce broad viral gene expression in monocytes In monocytes, immediate-early (IE) genes are repressed compared to all other HCMV genes
Collapse
Affiliation(s)
- Batsheva Rozman
- Department of Molecular Genetics, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Aharon Nachshon
- Department of Molecular Genetics, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Roi Levi Samia
- Department of Molecular Genetics, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Michael Lavi
- Department of Molecular Genetics, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Michal Schwartz
- Department of Molecular Genetics, Weizmann Institute of Science, 76100 Rehovot, Israel.
| | - Noam Stern-Ginossar
- Department of Molecular Genetics, Weizmann Institute of Science, 76100 Rehovot, Israel.
| |
Collapse
|
14
|
Uotani R, Miyazaki D, Shimizu Y, Ohtani F, Haruki T, Sasaki SI, Koyama A, Inoue Y, Suzutani T. Antiviral cytotoxic T lymphocyte responses for long term prognosis of corneal infection by cytomegalovirus in immunocompetent subjects. Sci Rep 2022; 12:5419. [PMID: 35354878 PMCID: PMC8967827 DOI: 10.1038/s41598-022-09312-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 02/28/2022] [Indexed: 11/09/2022] Open
Abstract
Ocular cytomegalovirus (CMV) infections in immunocompetent individuals are rare, but its activation can cause chronic and relapsing inflammation in anterior segment of the eye resulting in loss of corneal clarity and glaucoma. Fifty five patients with anterior segment CMV infection were assessed for their clinical characteristics, and CMV corneal endotheliitis was found to cause significant loss of corneal endothelial cells. The disease duration with recurrences was significantly correlated with the maximum intraocular level of CMV DNA. To examine why CMV is activated in healthy immunocompetent individuals and causing corneal endothelial cell damage, assays of cytotoxic T cells (CTLs) which directly target infected corneal endothelial cells were performed for 9 HLA-matched CMV corneal endotheliitis patients (HLA-A*2402). When the cell loss was analyzed for associations with CTL responses, CMV-induced endothelial cell damage was mitigated by pp65-specific CTL induction. The recurrence-free time was also prolonged by pp65-specific CTL induction (hazard ratio (HR): 0.93, P = 0.01). In contrast, IE1-specific CTL was associated with endothelial cell damage and reduced the time for corneal transplantation (HR: 1.6, P = 0.003) and glaucoma surgery (HR: 1.5, P = 0.001). Collectively, induction of pp65-specific CTL was associated with improved visual prognosis. However, IE1-specific CTL without proper induction of pp65-specific CTL can cause pathological damage leading to the need of surgical interventions.
Collapse
Affiliation(s)
- Ryu Uotani
- Division of Ophthalmology and Visual Science, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago Tottori, 683-8504, Japan
| | - Dai Miyazaki
- Division of Ophthalmology and Visual Science, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago Tottori, 683-8504, Japan.
| | - Yumiko Shimizu
- Division of Ophthalmology and Visual Science, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago Tottori, 683-8504, Japan
| | - Fumie Ohtani
- Division of Ophthalmology and Visual Science, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago Tottori, 683-8504, Japan
| | - Tomoko Haruki
- Division of Ophthalmology and Visual Science, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago Tottori, 683-8504, Japan
| | - Shin-Ichi Sasaki
- Division of Ophthalmology and Visual Science, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago Tottori, 683-8504, Japan
| | - Ayumi Koyama
- Division of Ophthalmology and Visual Science, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago Tottori, 683-8504, Japan
| | - Yoshitsugu Inoue
- Division of Ophthalmology and Visual Science, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago Tottori, 683-8504, Japan
| | | |
Collapse
|
15
|
Poloni C, Szyf M, Cheishvili D, Tsoukas CM. Are the Healthy Vulnerable? Cytomegalovirus Seropositivity in Healthy Adults Is Associated With Accelerated Epigenetic Age and Immune Dysregulation. J Infect Dis 2022; 225:443-452. [PMID: 34255838 PMCID: PMC8344607 DOI: 10.1093/infdis/jiab365] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 07/12/2021] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Evaluating age as a risk factor for susceptibility to infectious diseases, particularly coronavirus disease 2019 (COVID-19), is critical. Cytomegalovirus (CMV) serologic prevalence increases with age and associates with inflammatory-mediated diseases in the elderly. However, little is known regarding the subclinical impact of CMV and risk it poses to healthy older adults. Prior to the COVID-19 pandemic we conducted a study to determine the association of CMV to biologic age and immune dysregulation. METHODS Community-dwelling, healthy adults older than 60 years were evaluated using DNA methylation assays to define epigenetic age (EpiAge) and T-cell immunophenotyping to assess immune dysregulation. RESULTS All subjects were healthy and asymptomatic. Those CMV seropositive had more lymphocytes, CD8 T cells, CD28- T cells, decreased CD4:CD8 cell ratios, and had higher average EpiAge (65.34 years) than those CMV seronegative (59.53 years). Decreased percent CD4 (P = .003) and numbers of CD4 T cells (P = .0199) correlated with increased EpiAge. CONCLUSIONS Our novel findings distinguish altered immunity in the elderly based on CMV status. Chronic CMV infection in healthy, older adults is associated with indicators of immune dysregulation, both of which correlate to differences in EpiAge.
Collapse
Affiliation(s)
- Chad Poloni
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Moshe Szyf
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | | | - Christos M Tsoukas
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- Department of Medicine, Division of Allergy and Clinical Immunology, McGill University, Montreal, Quebec, Canada
- Division of Experimental Medicine, The Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
16
|
MCMV-based vaccine vectors expressing full-length viral proteins provide long-term humoral immune protection upon a single-shot vaccination. Cell Mol Immunol 2022; 19:234-244. [PMID: 34992275 PMCID: PMC8739032 DOI: 10.1038/s41423-021-00814-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 11/25/2021] [Indexed: 11/15/2022] Open
Abstract
Global pandemics caused by influenza or coronaviruses cause severe disruptions to public health and lead to high morbidity and mortality. There remains a medical need for vaccines against these pathogens. CMV (cytomegalovirus) is a β-herpesvirus that induces uniquely robust immune responses in which remarkably large populations of antigen-specific CD8+ T cells are maintained for a lifetime. Hence, CMV has been proposed and investigated as a novel vaccine vector for expressing antigenic peptides or proteins to elicit protective cellular immune responses against numerous pathogens. We generated two recombinant murine CMV (MCMV) vaccine vectors expressing hemagglutinin (HA) of influenza A virus (MCMVHA) or the spike protein of severe acute respiratory syndrome coronavirus 2 (MCMVS). A single injection of MCMVs expressing either viral protein induced potent neutralizing antibody responses, which strengthened over time. Importantly, MCMVHA-vaccinated mice were protected from illness following challenge with the influenza virus, and we excluded that this protection was due to the effects of memory T cells. Conclusively, we show here that MCMV vectors induce not only long-term cellular immunity but also humoral responses that provide long-term immune protection against clinically relevant respiratory pathogens.
Collapse
|
17
|
Host-Adapted Gene Families Involved in Murine Cytomegalovirus Immune Evasion. Viruses 2022; 14:v14010128. [PMID: 35062332 PMCID: PMC8781790 DOI: 10.3390/v14010128] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/08/2022] [Accepted: 01/10/2022] [Indexed: 12/12/2022] Open
Abstract
Cytomegaloviruses (CMVs) are host species-specific and have adapted to their respective mammalian hosts during co-evolution. Host-adaptation is reflected by “private genes” that have specialized in mediating virus-host interplay and have no sequence homologs in other CMV species, although biological convergence has led to analogous protein functions. They are mostly organized in gene families evolved by gene duplications and subsequent mutations. The host immune response to infection, both the innate and the adaptive immune response, is a driver of viral evolution, resulting in the acquisition of viral immune evasion proteins encoded by private gene families. As the analysis of the medically relevant human cytomegalovirus by clinical investigation in the infected human host cannot make use of designed virus and host mutagenesis, the mouse model based on murine cytomegalovirus (mCMV) has become a versatile animal model to study basic principles of in vivo virus-host interplay. Focusing on the immune evasion of the adaptive immune response by CD8+ T cells, we review here what is known about proteins of two private gene families of mCMV, the m02 and the m145 families, specifically the role of m04, m06, and m152 in viral antigen presentation during acute and latent infection.
Collapse
|
18
|
Ferri C, Arcangeletti MC, Caselli E, Zakrzewska K, Maccari C, Calderaro A, D'Accolti M, Soffritti I, Arvia R, Sighinolfi G, Artoni E, Giuggioli D. Insights into the knowledge of complex diseases: Environmental infectious/toxic agents as potential etiopathogenetic factors of systemic sclerosis. J Autoimmun 2021; 124:102727. [PMID: 34601207 DOI: 10.1016/j.jaut.2021.102727] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 12/14/2022]
Abstract
Systemic sclerosis (SSc) is a connective tissue disease secondary to three cardinal pathological features: immune-system alterations, diffuse microangiopathy, and fibrosis involving the skin and internal organs. The etiology of SSc remains quite obscure; it may encompass multiple host genetic and environmental -infectious/chemical-factors. The present review focused on the potential role of environmental agents in the etiopathogenesis of SSc based on epidemiological, clinical, and laboratory investigations previously published in the world literature. Among infectious agents, some viruses that may persist and reactivate in infected individuals, namely human cytomegalovirus (HCMV), human herpesvirus-6 (HHV-6), and parvovirus B19 (B19V), and retroviruses have been proposed as potential causative agents of SSc. These viruses share a number of biological activities and consequent pathological alterations, such as endothelial dysfunction and/or fibroblast activation. Moreover, the acute worsening of pre-existing interstitial lung involvement observed in SSc patients with symptomatic SARS-CoV-2 infection might suggest a potential role of this virus in the overall disease outcome. A variety of chemical/occupational agents might be regarded as putative etiological factors of SSc. In this setting, the SSc complicating silica dust exposure represents one of the most promising models of study. Considering the complexity of SSc pathogenesis, none of suggested causative factors may explain the appearance of the whole SSc; it is likely that the disease is the result of a multifactorial and multistep pathogenetic process. A variable combination of potential etiological factors may modulate the appearance of different clinical phenotypes detectable in individual scleroderma patients. The in-deep investigations on the SSc etiopathogenesis may provide useful insights in the broad field of human diseases characterized by diffuse microangiopathy or altered fibrogenesis.
Collapse
Affiliation(s)
- Clodoveo Ferri
- Rheumatology Unit, Medical School, University of Modena and Reggio E, University-Hospital Policlinico of Modena, Modena, Italy; Rheumatology Unit, Casa di Cura Madonna dello Scoglio, Cotronei (KR), Italy.
| | | | - Elisabetta Caselli
- Section of Microbiology, Department of Chemical, Pharmaceutical and Agricultural Sciences and LTTA, University of Ferrara, Ferrara, Italy
| | - Krystyna Zakrzewska
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Clara Maccari
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Adriana Calderaro
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Maria D'Accolti
- Section of Microbiology, Department of Chemical, Pharmaceutical and Agricultural Sciences and LTTA, University of Ferrara, Ferrara, Italy
| | - Irene Soffritti
- Section of Microbiology, Department of Chemical, Pharmaceutical and Agricultural Sciences and LTTA, University of Ferrara, Ferrara, Italy
| | - Rosaria Arvia
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Gianluca Sighinolfi
- Rheumatology Unit, Medical School, University of Modena and Reggio E, University-Hospital Policlinico of Modena, Modena, Italy.
| | - Erica Artoni
- Rheumatology Unit, Medical School, University of Modena and Reggio E, University-Hospital Policlinico of Modena, Modena, Italy
| | - Dilia Giuggioli
- Rheumatology Unit, Medical School, University of Modena and Reggio E, University-Hospital Policlinico of Modena, Modena, Italy
| |
Collapse
|
19
|
Blaum F, Lukas D, Reddehase MJ, Lemmermann NAW. Localization of Viral Epitope-Specific CD8 T Cells during Cytomegalovirus Latency in the Lungs and Recruitment to Lung Parenchyma by Airway Challenge Infection. Life (Basel) 2021; 11:life11090918. [PMID: 34575067 PMCID: PMC8467276 DOI: 10.3390/life11090918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 01/10/2023] Open
Abstract
Interstitial pneumonia is a life-threatening clinical manifestation of cytomegalovirus infection in recipients of hematopoietic cell transplantation (HCT). The mouse model of experimental HCT and infection with murine cytomegalovirus revealed that reconstitution of virus-specific CD8+ T cells is critical for resolving productive lung infection. CD8+ T-cell infiltrates persisted in the lungs after the establishment of latent infection. A subset defined by the phenotype KLRG1+CD62L− expanded over time, a phenomenon known as memory inflation (MI). Here we studied the localization of these inflationary T effector-memory cells (iTEM) by comparing their frequencies in the intravascular and transmigration compartments, the IVC and TMC, respectively, with their frequency in the extravascular compartment (EVC), the alveolar epithelium. Frequencies of viral epitope-specific iTEM were comparable in the IVC and TMC but were reduced in the EVC, corresponding to an increase in KLRG1−CD62L− conventional T effector-memory cells (cTEM) and a decrease in functional IFNγ+CD8+ T cells. As maintained expression of KLRG1 requires stimulation by antigen, we conclude that iTEM lose KLRG1 and convert to cTEM after transmigration into the EVC because pneumocytes are not latently infected and, therefore, do not express antigens. Accordingly, antigen re-expression upon airway challenge infection recruited virus-specific CD8+ T cells to TMC and EVC.
Collapse
Affiliation(s)
- Franziska Blaum
- Institute for Virology, Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (F.B.); (N.A.W.L.)
| | - Dominika Lukas
- Department of Dermatology, University of Cologne, University Hospital Cologne and Faculty of Medicine, 50937 Cologne, Germany;
| | - Matthias J. Reddehase
- Institute for Virology, Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (F.B.); (N.A.W.L.)
- Correspondence:
| | - Niels A. W. Lemmermann
- Institute for Virology, Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (F.B.); (N.A.W.L.)
| |
Collapse
|
20
|
Immunodominant Cytomegalovirus Epitopes Suppress Subdominant Epitopes in the Generation of High-Avidity CD8 T Cells. Pathogens 2021; 10:pathogens10080956. [PMID: 34451420 PMCID: PMC8400798 DOI: 10.3390/pathogens10080956] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/19/2021] [Accepted: 07/24/2021] [Indexed: 01/04/2023] Open
Abstract
CD8+ T-cell responses to pathogens are directed against infected cells that present pathogen-encoded peptides on MHC class-I molecules. Although natural responses are polyclonal, the spectrum of peptides that qualify for epitopes is remarkably small even for pathogens with high coding capacity. Among those few that are successful at all, a hierarchy exists in the magnitude of the response that they elicit in terms of numbers of CD8+ T cells generated. This led to a classification into immunodominant and non-immunodominant or subordinate epitopes, IDEs and non-IDEs, respectively. IDEs are favored in the design of vaccines and are chosen for CD8+ T-cell immunotherapy. Using murine cytomegalovirus as a model, we provide evidence to conclude that epitope hierarchy reflects competition on the level of antigen recognition. Notably, high-avidity cells specific for non-IDEs were found to expand only when IDEs were deleted. This may be a host’s back-up strategy to avoid viral immune escape through antigenic drift caused by IDE mutations. Importantly, our results are relevant for the design of vaccines based on cytomegaloviruses as vectors to generate high-avidity CD8+ T-cell memory specific for unrelated pathogens or tumors. We propose the deletion of vector-encoded IDEs to avoid the suppression of epitopes of the vaccine target.
Collapse
|
21
|
Direct Evidence for Viral Antigen Presentation during Latent Cytomegalovirus Infection. Pathogens 2021; 10:pathogens10060731. [PMID: 34200578 PMCID: PMC8229173 DOI: 10.3390/pathogens10060731] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/05/2021] [Accepted: 06/08/2021] [Indexed: 02/07/2023] Open
Abstract
Murine models of cytomegalovirus (CMV) infection have revealed an immunological phenomenon known as “memory inflation” (MI). After a peak of a primary CD8+ T-cell response, the pool of epitope-specific cells contracts in parallel to the resolution of productive infection and the establishment of a latent infection, referred to as “latency.” CMV latency is associated with an increase in the number of cells specific for certain viral epitopes over time. The inflationary subset was identified as effector-memory T cells (iTEM) characterized by the cell surface phenotype KLRG1+CD127−CD62L−. As we have shown recently, latent viral genomes are not transcriptionally silent. Rather, viral genes are sporadically desilenced in a stochastic fashion. The current hypothesis proposes MI to be driven by presented viral antigenic peptides encoded by the corresponding, stochastically expressed viral genes. Although this mechanism suggests itself, independent evidence for antigen presentation during viral latency is pending. Here we fill this gap by showing that T cell-receptor transgenic OT-I cells that are specific for peptide SIINFEKL proliferate upon adoptive cell transfer in C57BL/6 recipients latently infected with murine CMV encoding SIINFEKL (mCMV-SIINFEKL), but not in those latently infected with mCMV-SIINFEKA, in which antigenicity is lost by mutation L8A of the C-terminal amino acid residue.
Collapse
|
22
|
Griessl M, Renzaho A, Freitag K, Seckert CK, Reddehase MJ, Lemmermann NAW. Stochastic Episodes of Latent Cytomegalovirus Transcription Drive CD8 T-Cell "Memory Inflation" and Avoid Immune Evasion. Front Immunol 2021; 12:668885. [PMID: 33968074 PMCID: PMC8100209 DOI: 10.3389/fimmu.2021.668885] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/06/2021] [Indexed: 11/29/2022] Open
Abstract
Acute infection with murine cytomegalovirus (mCMV) is controlled by CD8+ T cells and develops into a state of latent infection, referred to as latency, which is defined by lifelong maintenance of viral genomes but absence of infectious virus in latently infected cell types. Latency is associated with an increase in numbers of viral epitope-specific CD8+ T cells over time, a phenomenon known as "memory inflation" (MI). The "inflationary" subset of CD8+ T cells has been phenotyped as KLRG1+CD62L- effector-memory T cells (iTEM). It is agreed upon that proliferation of iTEM requires repeated episodes of antigen presentation, which implies that antigen-encoding viral genes must be transcribed during latency. Evidence for this has been provided previously for the genes encoding the MI-driving antigenic peptides IE1-YPHFMPTNL and m164-AGPPRYSRI of mCMV in the H-2d haplotype. There exist two competing hypotheses for explaining MI-driving viral transcription. The "reactivation hypothesis" proposes frequent events of productive virus reactivation from latency. Reactivation involves a coordinated gene expression cascade from immediate-early (IE) to early (E) and late phase (L) transcripts, eventually leading to assembly and release of infectious virus. In contrast, the "stochastic transcription hypothesis" proposes that viral genes become transiently de-silenced in latent viral genomes in a stochastic fashion, not following the canonical IE-E-L temporal cascade of reactivation. The reactivation hypothesis, however, is incompatible with the finding that productive virus reactivation is exceedingly rare in immunocompetent mice and observed only under conditions of compromised immunity. In addition, the reactivation hypothesis fails to explain why immune evasion genes, which are regularly expressed during reactivation in the same cells in which epitope-encoding genes are expressed, do not prevent antigen presentation and thus MI. Here we show that IE, E, and L genes are transcribed during latency, though stochastically, not following the IE-E-L temporal cascade. Importantly, transcripts that encode MI-driving antigenic peptides rarely coincide with those that encode immune evasion proteins. As immune evasion can operate only in cis, that is, in a cell that simultaneously expresses antigenic peptides, the stochastic transcription hypothesis explains why immune evasion is not operative in latently infected cells and, therefore, does not interfere with MI.
Collapse
Affiliation(s)
| | | | | | | | | | - Niels A. W. Lemmermann
- Institute for Virology, Research Center for Immunotherapy (FZI) at the University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| |
Collapse
|
23
|
Abassi L, Cicin-Sain L. The avid competitors of memory inflation. Curr Opin Virol 2020; 44:162-168. [PMID: 33039898 DOI: 10.1016/j.coviro.2020.08.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/14/2020] [Accepted: 08/16/2020] [Indexed: 12/18/2022]
Abstract
Cytomegaloviruses (CMV) coevolve with their hosts and latently persist in the vast majority of adult mammals. Therefore, persistent T-cell responses to CMV antigens during virus latency offer a fascinating perspective on the evolution of the T-cell repertoire in natural settings. We addressed here the life-long interactions between CMV antigens presented on MHC-I molecules and the CD8 T-cell response. We present the mechanistic evidence from the murine model of CMV infection and put it in context of clinical laboratory results. We will highlight the remarkable parallels in T-cell responses between the two biological systems, and focus in particular on memory inflation as a result of competitive processes, both between viral antigenic peptides and between T-cell receptors on the host's cytotoxic lymphocytes.
Collapse
Affiliation(s)
- Leila Abassi
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Germany
| | - Luka Cicin-Sain
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School (MHH), Germany; Centre for Individualised Infection Medicine (CIIM), A Joint Venture of HZI and MHH, Germany; German Centre for Infection Research (DZIF), Hannover-Braunschweig Site, Germany.
| |
Collapse
|
24
|
Holtappels R, Freitag K, Renzaho A, Becker S, Lemmermann NA, Reddehase MJ. Revisiting CD8 T-cell 'Memory Inflation': New Insights with Implications for Cytomegaloviruses as Vaccine Vectors. Vaccines (Basel) 2020; 8:vaccines8030402. [PMID: 32707744 PMCID: PMC7563500 DOI: 10.3390/vaccines8030402] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/09/2020] [Accepted: 07/17/2020] [Indexed: 12/12/2022] Open
Abstract
Murine models of cytomegalovirus (CMV) infection have revealed an exceptional kinetics of the immune response. After resolution of productive infection, transient contraction of the viral epitope-specific CD8 T-cell pool was found to be followed by a pool expansion specific for certain viral epitopes during non-productive ‘latent’ infection. This phenomenon, known as ‘memory inflation’ (MI), was found to be based on inflationary KLRG1+CD62L− effector-memory T cells (iTEM) that depend on repetitive restimulation. MI gained substantial interest for employing CMV as vaccine vector by replacing MI-driving CMV epitopes with foreign epitopes for generating high numbers of protective memory cells specific for unrelated pathogens. The concept of an MI-driving CMV vector is questioned by human studies disputing MI in humans. A bias towards MI in experimental models may have resulted from systemic infection. We have here studied local murine CMV infection as a route that is more closely matching routine human vaccine application. Notably, KLRG1−CD62L+ central memory T cells (TCM) and conventional KLRG1−CD62L− effector memory T cells (cTEM) were found to expand, associated with ‘avidity maturation’, whereas the pool size of iTEM steadily declined over time. The establishment of high avidity CD8 T-cell central memory encourages one to pursue the concept of CMV vector-based vaccines.
Collapse
|
25
|
Sex-Differential Impact of Human Cytomegalovirus Infection on In Vitro Reactivity to Toll-Like Receptor 2, 4 and 7/8 Stimulation in Gambian Infants. Vaccines (Basel) 2020; 8:vaccines8030407. [PMID: 32707906 PMCID: PMC7564534 DOI: 10.3390/vaccines8030407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/14/2020] [Accepted: 07/20/2020] [Indexed: 11/17/2022] Open
Abstract
Human cytomegalovirus (HCMV) infection rates approach 100% by the first year of life in low-income countries. It is not known if this drives changes to innate immunity in early life and thereby altered immune reactivity to infections and vaccines. Given the panoply of sex differences in immunity, it is feasible that any immunological effects of HCMV would differ in males and females. We analysed ex vivo innate cytokine responses to a panel of toll-like receptor (TLR) ligands in 108 nine-month-old Gambian males and females participating in a vaccine trial. We found evidence that HCMV suppressed reactivity to TLR2 and TLR7/8 stimulation in females but not males. This is likely to contribute to sex differences in responses to infections and vaccines in early life and has implications for the development of TLR ligands as vaccine adjuvants. Development of an effective HCMV vaccine would be able to circumvent some of these potentially negative effects of HCMV infection in childhood.
Collapse
|
26
|
|
27
|
Smith CJ, Venturi V, Quigley MF, Turula H, Gostick E, Ladell K, Hill BJ, Himelfarb D, Quinn KM, Greenaway HY, Dang THY, Seder RA, Douek DC, Hill AB, Davenport MP, Price DA, Snyder CM. Stochastic Expansions Maintain the Clonal Stability of CD8 + T Cell Populations Undergoing Memory Inflation Driven by Murine Cytomegalovirus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:112-121. [PMID: 31818981 PMCID: PMC6920548 DOI: 10.4049/jimmunol.1900455] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 10/16/2019] [Indexed: 11/19/2022]
Abstract
CMV is an obligate and persistent intracellular pathogen that continually drives the production of highly differentiated virus-specific CD8+ T cells in an Ag-dependent manner, a phenomenon known as memory inflation. Extensive proliferation is required to generate and maintain inflationary CD8+ T cell populations, which are counterintuitively short-lived and typically exposed to limited amounts of Ag during the chronic phase of infection. An apparent discrepancy therefore exists between the magnitude of expansion and the requirement for ongoing immunogenic stimulation. To address this issue, we explored the clonal dynamics of memory inflation. First, we tracked congenically marked OT-I cell populations in recipient mice infected with murine CMV (MCMV) expressing the cognate Ag OVA. Irrespective of numerical dominance, stochastic expansions were observed in each population, such that dominant and subdominant OT-I cells were maintained at stable frequencies over time. Second, we characterized endogenous CD8+ T cell populations specific for two classic inflationary epitopes, M38 and IE3. Multiple clonotypes simultaneously underwent Ag-driven proliferation during latent infection with MCMV. In addition, the corresponding CD8+ T cell repertoires were stable over time and dominated by persistent clonotypes, many of which also occurred in more than one mouse. Collectively, these data suggest that stochastic encounters with Ag occur frequently enough to maintain oligoclonal populations of inflationary CD8+ T cells, despite intrinsic constraints on epitope display at individual sites of infection with MCMV.
Collapse
Affiliation(s)
- Corinne J Smith
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Vanessa Venturi
- Infection Analytics Program, Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, New South Wales 2052, Australia
| | - Maire F Quigley
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Holly Turula
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Emma Gostick
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff CF14 4XN, United Kingdom
| | - Kristin Ladell
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff CF14 4XN, United Kingdom
| | - Brenna J Hill
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Danielle Himelfarb
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Kylie M Quinn
- Cellular Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| | - Hui Yee Greenaway
- Infection Analytics Program, Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, New South Wales 2052, Australia
| | - Thurston H Y Dang
- Infection Analytics Program, Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, New South Wales 2052, Australia
| | - Robert A Seder
- Cellular Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| | - Daniel C Douek
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Ann B Hill
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239
| | - Miles P Davenport
- Infection Analytics Program, Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, New South Wales 2052, Australia
| | - David A Price
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892;
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff CF14 4XN, United Kingdom
| | - Christopher M Snyder
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107;
| |
Collapse
|
28
|
Dons’koi BV, Tutchenko TM, Chernyshov VP, Stepaniuk KS. HCMV seropositivity is associated with specific proinflammatory immune phenotype in women with implantation failure. Immunol Lett 2020; 217:84-90. [DOI: 10.1016/j.imlet.2019.11.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 11/07/2019] [Accepted: 11/17/2019] [Indexed: 01/06/2023]
|
29
|
Vaccine Vectors Harnessing the Power of Cytomegaloviruses. Vaccines (Basel) 2019; 7:vaccines7040152. [PMID: 31627457 PMCID: PMC6963789 DOI: 10.3390/vaccines7040152] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/11/2019] [Accepted: 10/14/2019] [Indexed: 02/05/2023] Open
Abstract
Cytomegalovirus (CMV) species have been gaining attention as experimental vaccine vectors inducing cellular immune responses of unparalleled strength and protection. This review outline the strengths and the restrictions of CMV-based vectors, in light of the known aspects of CMV infection, pathogenicity and immunity. We discuss aspects to be considered when optimizing CMV based vaccines, including the innate immune response, the adaptive humoral immunity and the T-cell responses. We also discuss the antigenic epitopes presented by unconventional major histocompatibility complex (MHC) molecules in some CMV delivery systems and considerations about routes for delivery for the induction of systemic or mucosal immune responses. With the first clinical trials initiating, CMV-based vaccine vectors are entering a mature phase of development. This impetus needs to be maintained by scientific advances that feed the progress of this technological platform.
Collapse
|
30
|
Immunization with a murine cytomegalovirus based vector encoding retrovirus envelope confers strong protection from Friend retrovirus challenge infection. PLoS Pathog 2019; 15:e1008043. [PMID: 31568492 PMCID: PMC6786657 DOI: 10.1371/journal.ppat.1008043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 10/10/2019] [Accepted: 08/25/2019] [Indexed: 02/04/2023] Open
Abstract
Immunization vectors based on cytomegalovirus (CMV) have attracted a lot of interest in recent years because of their high efficacy in the simian immunodeficiency virus (SIV) macaque model, which has been attributed to their ability to induce strong, unusually broad, and unconventionally restricted CD8+ T cell responses. To evaluate the ability of CMV-based vectors to mediate protection by other immune mechanisms, we evaluated a mouse CMV (MCMV)-based vector encoding Friend virus (FV) envelope (Env), which lacks any known CD8+ T cell epitopes, for its protective efficacy in the FV mouse model. When we immunized highly FV-susceptible mice with the Env-encoding MCMV vector (MCMV.env), we could detect high frequencies of Env-specific CD4+ T cells after a single immunization. While the control of an early FV challenge infection was highly variable, an FV infection applied later after immunization was tightly controlled by almost all immunized mice. Protection of mice correlated with their ability to mount a robust anamnestic neutralizing antibody response upon FV infection, but Env-specific CD4+ T cells also produced appreciable levels of interferon γ. Depletion and transfer experiments underlined the important role of antibodies for control of FV infection but also showed that while no Env-specific CD8+ T cells were induced by the MCMV.env vaccine, the presence of CD8+ T cells at the time of FV challenge was required. The immunity induced by MCMV.env immunization was long-lasting, but was restricted to MCMV naïve animals. Taken together, our results demonstrate a novel mode of action of a CMV-based vaccine for anti-retrovirus immunization that confers strong protection from retrovirus challenge, which is conferred by CD4+ T cells and antibodies. CMV-based vectors have attracted a lot of attention in the vaccine development field, since they were shown to induce unconventionally restricted CD8+ T cell responses and strong protection in the SIV rhesus macaque model. In a mouse retrovirus model, we show now that immunization with a mouse CMV-based vector encoding retrovirus envelope conferred very strong protection, even though it was not designed to induce any CD8+ T cell responses. In this MCMV.env immunization, protection relied on the induction of CD4+ T cells and the ability to mount a strong anamnestic neutralizing antibody response upon retrovirus infection, but it was restricted to MCMV pre-naïve mice. In our model system, the MCMV based vector shows very high efficacy that is comparable to an attenuated retrovirus-based vaccine, and encourages the pursuit of this vaccination strategy.
Collapse
|
31
|
van den Berg SPH, Pardieck IN, Lanfermeijer J, Sauce D, Klenerman P, van Baarle D, Arens R. The hallmarks of CMV-specific CD8 T-cell differentiation. Med Microbiol Immunol 2019; 208:365-373. [PMID: 30989333 PMCID: PMC6647465 DOI: 10.1007/s00430-019-00608-7] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 04/02/2019] [Indexed: 12/13/2022]
Abstract
Upon cytomegalovirus (CMV) infection, large T-cell responses are elicited that remain high or even increase over time, a phenomenon named memory T-cell inflation. Besides, the maintained robust T-cell response, CMV-specific T cells seem to have a distinctive phenotype, characterized by an advanced differentiation state. Here, we will review this "special" differentiation status by discussing the cellular phenotype based on the expression of CD45 isoforms, costimulatory, inhibitory and natural killer receptors, adhesion and lymphocyte homing molecules, transcription factors, cytokines and cytotoxic molecules. In addition, we focus on whether the differentiation state of CMV-specific CD8 T cells is unique in comparison with other chronic viruses and we will discuss the possible impact of factors such as antigen exposure and aging on the advanced differentiation status of CMV-specific CD8 T cells.
Collapse
Affiliation(s)
- Sara P H van den Berg
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
- Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Iris N Pardieck
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Josien Lanfermeijer
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
- Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Delphine Sauce
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Paul Klenerman
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
- NIHR Biomedical Research Centre, John Radcliffe Hospital, Oxford, UK
| | - Debbie van Baarle
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
- Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ramon Arens
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
32
|
Krmpotić A, Podlech J, Reddehase MJ, Britt WJ, Jonjić S. Role of antibodies in confining cytomegalovirus after reactivation from latency: three decades' résumé. Med Microbiol Immunol 2019; 208:415-429. [PMID: 30923898 PMCID: PMC6705608 DOI: 10.1007/s00430-019-00600-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 03/18/2019] [Indexed: 12/16/2022]
Abstract
Cytomegaloviruses (CMVs) are highly prevalent herpesviruses, characterized by strict species specificity and the ability to establish non-productive latent infection from which reactivation can occur. Reactivation of latent human CMV (HCMV) represents one of the most important clinical challenges in transplant recipients secondary to the strong immunosuppression. In addition, HCMV is the major viral cause of congenital infection with severe sequelae including brain damage. The accumulated evidence clearly shows that cellular immunity plays a major role in the control of primary CMV infection as well as establishment and maintenance of latency. However, the efficiency of antiviral antibodies in virus control, particularly in prevention of congenital infection and virus reactivation from latency in immunosuppressed hosts, is much less understood. Because of a strict species specificity of HCMV, the role of antibodies in controlling CMV disease has been addressed using murine CMV (MCMV) as a model. Here, we review and discuss the role played by the antiviral antibody response during CMV infections with emphasis on latency and reactivation not only in the MCMV model, but also in relevant clinical settings. We provide evidence to conclude that antiviral antibodies do not prevent the initiating molecular event of virus reactivation from latency but operate by preventing intra-organ spread and inter-organ dissemination of recurrent virus.
Collapse
Affiliation(s)
- Astrid Krmpotić
- Department of Histology and Embryology and Center for Proteomics, University of Rijeka, Faculty of Medicine, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Jürgen Podlech
- Institute for Virology and Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Strasse 67, 55131, Mainz, Germany
| | - Matthias J. Reddehase
- Institute for Virology and Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Strasse 67, 55131, Mainz, Germany
| | - William J. Britt
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA and Department of Pediatrics Infectious Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stipan Jonjić
- Department of Histology and Embryology and Center for Proteomics, University of Rijeka, Faculty of Medicine, Braće Branchetta 20, 51000 Rijeka, Croatia
| |
Collapse
|
33
|
Jergović M, Contreras NA, Nikolich-Žugich J. Impact of CMV upon immune aging: facts and fiction. Med Microbiol Immunol 2019; 208:263-269. [PMID: 31004198 PMCID: PMC6635032 DOI: 10.1007/s00430-019-00605-w] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 03/30/2019] [Indexed: 12/28/2022]
Abstract
Aging is accompanied by significant defects in immunity and compromised responses to new, previously unencountered microbial pathogens. Most humans carry several persistent or latent viruses as they age, interacting with the host immune systems for years. In that context maybe the most studied persistent virus is Cytomegalovirus, infamous for its ability to recruit very large T cell responses which increase with age and to simultaneously evade elimination by the immune system. Here we will address how lifelong CMV infection and the immunological burden of its control might affect immune reactivity and health of the host over time.
Collapse
Affiliation(s)
- Mladen Jergović
- Department of Immunobiology and the University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85718, USA
| | - Nico A Contreras
- Department of Immunobiology and the University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85718, USA
| | - Janko Nikolich-Žugich
- Department of Immunobiology and the University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85718, USA.
- University of Arizona College of Medicine-Tucson, 1501 N Campbell Ave, P.O. Box 221245, Tucson, AZ, 85724, USA.
| |
Collapse
|
34
|
Gabel M, Baumann NS, Oxenius A, Graw F. Investigating the Dynamics of MCMV-Specific CD8 + T Cell Responses in Individual Hosts. Front Immunol 2019; 10:1358. [PMID: 31281313 PMCID: PMC6595046 DOI: 10.3389/fimmu.2019.01358] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 05/29/2019] [Indexed: 01/03/2023] Open
Abstract
Infection by Cytomegalovirus (CMV) is characterized by the massive expansion and continued maintenance of CMV-specific CD8+ T cells for certain CMV-derived peptides. This phenomenon called “memory inflation" has made CMV a primary target for the generation of T cell based vaccine vectors against various diseases. However, many aspects concerning the generation and maintenance of the inflationary CD8+ T cell response still remain to be resolved. In this study, we combined experimental data and mathematical models to analyze the dynamics of circulatory inflationary CD8+ T cells within individual mice infected by MCMV. Obtaining frequent measurements on the number and frequency of CMV-specific CD8+ T cells up to 70 days post infection, we find that mathematical models assuming differing viral stimuli during acute infection and the inflationary phase provide a better description for the observed dynamics than models relying on similar viral stimuli during both phases. In addition, our analysis allowed a detailed quantification of the different phases of memory inflation within individual mice (1st-expansion - contraction - 2nd expansion/maintenance) indicating remarkable consistency of the timing of these phases across mice, but considerable variation in the size of the individual responses between mice. Our analysis provides a first step toward generating a mechanistic framework for analyzing the generation and maintenance of inflationary CD8+ T cells while accounting for individual heterogeneity. Extending these analyses by incorporating measurements from additional compartments and more prolonged sampling will help to obtain a systematic and quantitative understanding of the factors regulating the process of memory inflation.
Collapse
Affiliation(s)
- Michael Gabel
- Center for Modelling and Simulation in the Biosciences, BioQuant-Center, Heidelberg University, Heidelberg, Germany
| | - Nicolas S Baumann
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Annette Oxenius
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Frederik Graw
- Center for Modelling and Simulation in the Biosciences, BioQuant-Center, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
35
|
Reddehase MJ. 'Checks and balances' in cytomegalovirus-host cohabitation. Med Microbiol Immunol 2019; 208:259-261. [PMID: 31129788 DOI: 10.1007/s00430-019-00618-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 05/05/2019] [Indexed: 01/09/2023]
Affiliation(s)
- Matthias J Reddehase
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Strasse 67, 55131, Mainz, Germany.
| |
Collapse
|
36
|
Adler SP, Reddehase MJ. Pediatric roots of cytomegalovirus recurrence and memory inflation in the elderly. Med Microbiol Immunol 2019; 208:323-328. [PMID: 31062089 DOI: 10.1007/s00430-019-00609-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 04/05/2019] [Indexed: 12/11/2022]
Abstract
The establishment of a lifelong latent infection after resolution of primary infection is a hallmark of cytomegalovirus (CMV) biology. Primary infection with human CMV is possible any time in life, but most frequently, virus transmission occurs already perinatally or in early childhood. Many years or even decades later, severe clinical problems can result from recurrence of infectious virus by reactivation from latency in individuals who undergo immunocompromising medical treatment, for instance, transplant recipients, but also in septic patients without canonical immunosuppression, and in elderly people with a weakened immune system. The diversity of disease manifestations, such as retinitis, pneumonia, hepatitis, gastrointestinal disease, and others, has remained an enigma. In clinical routine, seropositivity for IgG antibodies against human CMV is taken to indicate latent infection and thus to define a qualitative risk of recurrence, but it is insufficient as a predictor for the quantitative risk of recurrence. Early experimental studies in the mouse model, comparing primary infection of neonatal and adult mice, led to the hypothesis that high load of latent viral genomes is a better predictor for the quantitative risk. A prolonged period of virus multiplication in the immunologically immature neonatally infected host increased the risk of virus recurrence by an enhanced copy number of latent virus genomes from which reactivation can initiate. In extension of this hypothesis, one would predict today that a higher incidence of reactivation events will also fuel the expansion of virus-specific T cells observed in the elderly, a phenomenon known as "memory inflation". Notably, the mouse model also indicated a stochastic nature of reactivation, thus offering an explanation for the diversity and organ selectivity of disease manifestations observed in patients. As the infection history is mostly undefined in humans, such predictions from the mouse model are difficult to verify by clinical investigation, and moreover, such questions were actually rarely addressed. Here, we have surveyed the existing literature for reports that may help to retrospectively relate the individual infection history to the risk of virus recurrence and recrudescent organ disease.
Collapse
Affiliation(s)
- Stuart P Adler
- CMV Research Foundation, 9304 Bandock Road, Richmond, VA, 23229, USA.
| | - Matthias J Reddehase
- Institute for Virology and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Strasse 67, Hochhaus am Augustusplatz, 55131, Mainz, Germany.
| |
Collapse
|
37
|
Reddehase MJ, Lemmermann NAW. Cellular reservoirs of latent cytomegaloviruses. Med Microbiol Immunol 2019; 208:391-403. [PMID: 31011793 DOI: 10.1007/s00430-019-00592-y] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 03/07/2019] [Indexed: 12/14/2022]
Abstract
Cytomegaloviruses (CMVs), members of the β-subfamily of the herpesvirus family, have co-speciated with their respective mammalian hosts resulting in a mutual virus-host adaptation reflected by sets of 'private' viral genes that a particular CMV species does not share with other CMVs and that define the host-species specificity of CMVs. Nonetheless, based on "biological convergence" in evolution, fundamental rules in viral pathogenesis and immune control are functionally analogous between different virus-host pairs. Therefore, the mouse model of infection with murine CMV (mCMV) has revealed generally valid principles of CMV-host interactions. Specifically, the mouse model has paved the way to cellular immunotherapy of CMV disease in immunocompromised recipients of hematopoietic cell transplantation (HCT). Precisely in the context of HCT, however, current view assumes that there exists a major difference between hCMV and mCMV regarding "latent virus reservoirs" in that only hCMV establishes latency in hematopoietic lineage cells (HLCs), whereas mCMV establishes latency in endothelial cells. This would imply that only hCMV can reactivate from transplanted HLCs of a latently infected donor. In addition, as viral transcriptional activity during latency is discussed as a driver of clonal T-cell expansion over lifetime, a phenomenon known as "memory inflation", it is important to know if hCMV and mCMV establish latency in the same cell type(s) for imprinting the immune system. Here, we review the currently available evidence to propose that the alleged difference in latent virus reservoirs between hCMV and mCMV may rather relate to a difference in the focus of research. While studies on hCMV latency in HLCs likely described a non-canonical, transient type-2 latency, studies in the mouse model focussed on canonical, lifelong type-1 latency.
Collapse
Affiliation(s)
- Matthias J Reddehase
- Institute for Virology and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Strasse 67, Hochhaus am Augustusplatz, 55131, Mainz, Germany.
| | - Niels A W Lemmermann
- Institute for Virology and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Strasse 67, Hochhaus am Augustusplatz, 55131, Mainz, Germany.
| |
Collapse
|
38
|
Transcripts expressed in cytomegalovirus latency coding for an antigenic IE/E phase peptide that drives "memory inflation". Med Microbiol Immunol 2019; 208:439-446. [PMID: 31004200 DOI: 10.1007/s00430-019-00615-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 04/11/2019] [Indexed: 01/01/2023]
Abstract
Roizman's definition of herpesviral latency, which applies also to cytomegaloviruses (CMVs), demands maintenance of reactivation-competent viral genomes after clearance of productive infection. It is more recent understanding that failure to complete the productive viral cycle for virus assembly and release does not imply viral gene silencing at all genetic loci and all the time. It rather appears that CMV latency is transcriptionally "noisy" in that silenced viral genes get desilenced from time to time in a stochastic manner, leading to "transcripts expressed in latency" (TELs). If a TEL happens to code for a protein that contains a CD8 T cell epitope, protein processing can lead to the presentation of the antigenic peptide and restimulation of cognate CD8 T cells during latency. This mechanism is discussed as a potential driver of epitope-selective accumulation of CD8 T cells over time, a phenomenon linked to CMV latency and known as "memory inflation" (MI). So far, expression of an epitope-encoding TEL was shown only for the major immediate-early (MIE) gene m123/ie1 of murine cytomegalovirus (mCMV), which codes for the prototypic MI-driving antigenic peptide YPHFMPTNL that is presented by the MHC class-I molecule Ld. The only known second MI-driving antigenic peptide of mCMV in the murine MHC haplotype H-2d is AGPPRYSRI presented by the MHC-I molecule Dd. This peptide is very special in that it is encoded by the early (E) phase gene m164 and by an overlapping immediate-early (IE) transcript governed by a promoter upstream of m164. If MI is driven by presentation of TEL-derived antigenic peptides, as the hypothesis says, one should find corresponding TELs. We show here that E-phase and IE-phase transcripts that code for the MI-driving antigenic peptide AGPPRYSRI are independently and stochastically expressed in latently infected lungs.
Collapse
|
39
|
Marandu T, Dombek M, Cook CH. Impact of cytomegalovirus load on host response to sepsis. Med Microbiol Immunol 2019; 208:295-303. [DOI: 10.1007/s00430-019-00603-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 03/22/2019] [Indexed: 12/24/2022]
|
40
|
Cicin-Sain L. Cytomegalovirus memory inflation and immune protection. Med Microbiol Immunol 2019; 208:339-347. [PMID: 30972476 DOI: 10.1007/s00430-019-00607-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 04/01/2019] [Indexed: 10/27/2022]
Abstract
Cytomegalovirus (CMV) infection induces powerful and sustained T-cell responses against a few selected immunodominant antigenic epitopes. This immune response was named memory inflation, because it does not contract in the long term, and may even expand over months and years of virus latency. It is by now understood that memory inflation does not occur at the expense of the naïve T-cell pool, but rather as a competitive selection process within the effector pool, where viral antigens with higher avidity of TCR binding and with earlier expression patterns outcompete those that are expressed later and bind TCRs less efficiently. It is also understood that inflationary epitopes require processing by the constitutive proteasome in non-hematopoietic cells, and this likely implies that memory inflation is fuelled by direct low-level antigenic expression in latently infected cells. This review proposes that these conditions make inflationary epitopes the optimal candidates for adoptive immunotherapy of CMV disease in the immunocompromised host. At present, functional target CMV epitopes have been defined only for the most common HLA haplotypes. Mapping the uncharacterized inflationary epitopes in less frequent HLAs may, thus, be a strategy for the identification of optimal immunotherapeutic targets in patients with uncommon haplotypes.
Collapse
Affiliation(s)
- Luka Cicin-Sain
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany. .,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School (MHH), Hannover, Germany. .,Centre for Individualised Infection Medicine (CIIM), A Joint Venture of HZI and MHH, Braunschweig, Germany. .,German Centre for Infection Research (DZIF), Hannover-Braunschweig site, Braunschweig, Germany.
| |
Collapse
|
41
|
Picarda G, Benedict CA. Cytomegalovirus: Shape-Shifting the Immune System. THE JOURNAL OF IMMUNOLOGY 2019; 200:3881-3889. [PMID: 29866770 DOI: 10.4049/jimmunol.1800171] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 03/26/2018] [Indexed: 11/19/2022]
Abstract
Systems-based based approaches have begun to shed light on extrinsic factors that contribute to immune system variation. Among these, CMV (HHV-5, a β-herpesvirus) imposes a surprisingly profound impact. Most of the world's population is CMV+, and the virus goes through three distinct infection phases en route to establishing lifelong détente with its host. Immune control of CMV in each phase recruits unique arms of host defense, and in turn the virus employs multiple immune-modulatory strategies that help facilitate the establishment of lifelong persistence. In this review, we explain how CMV shapes immunity and discuss the impact it may have on overall health.
Collapse
Affiliation(s)
- Gaëlle Picarda
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; and
| | - Chris A Benedict
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; and .,Center for Infectious Disease, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| |
Collapse
|
42
|
Cytomegalovirus-specific CD8+ T-cells are associated with a reduced incidence of early relapse after allogeneic stem cell transplantation. PLoS One 2019; 14:e0213739. [PMID: 30889204 PMCID: PMC6424430 DOI: 10.1371/journal.pone.0213739] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 02/27/2019] [Indexed: 01/05/2023] Open
Abstract
Leukemia relapse is the main cause for mortality after allogeneic stem cell transplantation (allo-SCT). Donor-derived allo-immune responses eliminate the residual host hematopoiesis and protect against relapse. Cytomegalovirus (CMV) reactivation (CMV-R) after allo-SCT may trigger anti-leukemic effects. The impact of CMV-specific CD8+ T-cells (CMV-CTLs) on the outcome after allo-SCT is currently unknown. Here, we studied the relationship between CMV-CTLs, overall T-cell reconstitution and relapse incidence in 103 patients with acute leukemia (n = 91) or myelodysplastic syndrome (n = 12) following CMV-seropositive recipient/donor (R+/D+) allo-SCT. Patients were subdivided based on the presence or absence of CMV-CTLs at 3 months after allo-SCT. Presence of CMV-CTLs was associated with preceding CMV-R and a fast T-cell reconstitution. Univariate analysis showed a significantly lower 1-, 2- and 5-year cumulative incidence of relapse (CIR) in patients with CMV-CTLs compared to those without CMV-CTLs. Multivariable regression analysis of the outcome performed with other relevant parameters chosen from univariate analysis revealed that presence of CMV-CTLs and chronic graft-versus-host disease (cGvHD) were the only independent factors associated with a low CIR. Onset of relapse was significantly later in patients with CMV-CTLs (median 489 days) than in in those without (median 152 days, p = 0.041) during a five-year follow-up. Presence of CMV-CTLs was associated with a lower incidence of early relapses (1 and 2-years), while cGvHD lead to a lower incidence of late relapses (2 to 5-years). In conclusion, our data show that CMV-CTLs indicate a functional immune-reconstitution protective against early relapse.
Collapse
|
43
|
Abstract
Human cytomegalovirus (HCMV) latency and reactivation is regulated by the chromatin structure at the major immediate early promoter (MIEP) within myeloid cells. Both cellular and viral factors are known to control this promoter during latency, here we will review the known mechanisms for MIEP regulation during latency. We will then focus on the virally encoded G-protein coupled receptor, US28, which suppresses the MIEP in early myeloid lineage cells. The importance of this function is underlined by the fact that US28 is essential for HCMV latency in CD34+ progenitor cells and CD14+ monocytes. We will describe cellular signalling pathways modulated by US28 to direct MIEP suppression during latency and demonstrate how US28 is able to ‘regulate the regulators’ of HCMV latency. Finally, we will describe how cell-surface US28 can be a target for antiviral therapies directed at the latent viral reservoir.
Collapse
|
44
|
Reddehase MJ, Lemmermann NAW. Mouse Model of Cytomegalovirus Disease and Immunotherapy in the Immunocompromised Host: Predictions for Medical Translation that Survived the "Test of Time". Viruses 2018; 10:v10120693. [PMID: 30563202 PMCID: PMC6315540 DOI: 10.3390/v10120693] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 11/26/2018] [Accepted: 12/03/2018] [Indexed: 12/14/2022] Open
Abstract
Human Cytomegalovirus (hCMV), which is the prototype member of the β-subfamily of the herpesvirus family, is a pathogen of high clinical relevance in recipients of hematopoietic cell transplantation (HCT). hCMV causes multiple-organ disease and interstitial pneumonia in particular upon infection during the immunocompromised period before hematopoietic reconstitution restores antiviral immunity. Clinical investigation of pathomechanisms and of strategies for an immune intervention aimed at restoring antiviral immunity earlier than by hematopoietic reconstitution are limited in patients to observational studies mainly because of ethical issues including the imperative medical indication for chemotherapy with antivirals. Aimed experimental studies into mechanisms, thus, require animal models that match the human disease as close as possible. Any model for hCMV disease is, however, constrained by the strict host-species specificity of CMVs that prevents the study of hCMV in any animal model including non-human primates. During eons of co-speciation, CMVs each have evolved a set of "private genes" in adaptation to their specific mammalian host including genes that have no homolog in the CMV virus species of any other host species. With a focus on the mouse model of CD8 T cell-based immunotherapy of CMV disease after experimental HCT and infection with murine CMV (mCMV), we review data in support of the phenomenon of "biological convergence" in virus-host adaptation. This includes shared fundamental principles of immune control and immune evasion, which allows us to at least make reasoned predictions from the animal model as an experimental "proof of concept." The aim of a model primarily is to define questions to be addressed by clinical investigation for verification, falsification, or modification and the results can then give feedback to refine the experimental model for research from "bedside to bench".
Collapse
Affiliation(s)
- Matthias J Reddehase
- Institute for Virology, University Medical Center and Center for Immunotherapy of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany.
| | - Niels A W Lemmermann
- Institute for Virology, University Medical Center and Center for Immunotherapy of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany.
| |
Collapse
|
45
|
Arcangeletti MC, Maccari C, Vescovini R, Volpi R, Giuggioli D, Sighinolfi G, De Conto F, Chezzi C, Calderaro A, Ferri C. A Paradigmatic Interplay between Human Cytomegalovirus and Host Immune System: Possible Involvement of Viral Antigen-Driven CD8+ T Cell Responses in Systemic Sclerosis. Viruses 2018; 10:E508. [PMID: 30231575 PMCID: PMC6163388 DOI: 10.3390/v10090508] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/12/2018] [Accepted: 09/14/2018] [Indexed: 12/12/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a highly prevalent opportunistic agent in the world population, which persists as a latent virus after a primary infection. Besides the well-established role of this agent causing severe diseases in immunocompromised individuals, more recently, HCMV has been evoked as a possible factor contributing to the pathogenesis of autoimmune diseases such as systemic sclerosis (SSc). The interplay between HCMV and immune surveillance is supposed to become unbalanced in SSc patients with expanded anti-HCMV immune responses, which are likely involved in the exacerbation of inflammatory processes. In this study, blood samples from a cohort of SSc patients vs. healthy subjects were tested for anti-HCMV immune responses (IgM, IgG antibodies, and T cells to peptide pools spanning the most immunogenic HCMV proteins). Statistically significant increase of HCMV-specific CD8+ T cell responses in SSc patients vs. healthy subjects was observed. Moreover, significantly greater HCMV-specific CD8+ T cell responses were found in SSc patients with a longer disease duration and those with higher modified Rodnan skin scores. Given the known importance of T cells in the development of SSc and that this virus may contribute to chronic inflammatory diseases, these data support a relevant role of HCMV-specific CD8+ T cell responses in SSc pathogenesis.
Collapse
Affiliation(s)
- Maria-Cristina Arcangeletti
- Virology Unit, University-Hospital of Parma, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy.
| | - Clara Maccari
- Virology Unit, University-Hospital of Parma, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy.
| | - Rosanna Vescovini
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy.
| | - Riccardo Volpi
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy.
| | - Dilia Giuggioli
- Rheumatology Unit, Medical School, University of Modena and Reggio Emilia, University-Hospital Policlinico of Modena, 41121 Modena, Italy.
| | - Gianluca Sighinolfi
- Rheumatology Unit, Medical School, University of Modena and Reggio Emilia, University-Hospital Policlinico of Modena, 41121 Modena, Italy.
| | - Flora De Conto
- Virology Unit, University-Hospital of Parma, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy.
| | - Carlo Chezzi
- Virology Unit, University-Hospital of Parma, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy.
| | - Adriana Calderaro
- Virology Unit, University-Hospital of Parma, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy.
| | - Clodoveo Ferri
- Rheumatology Unit, Medical School, University of Modena and Reggio Emilia, University-Hospital Policlinico of Modena, 41121 Modena, Italy.
| |
Collapse
|
46
|
Morabito KM, Ruckwardt TJ, Bar-Haim E, Nair D, Moin SM, Redwood AJ, Price DA, Graham BS. Memory Inflation Drives Tissue-Resident Memory CD8 + T Cell Maintenance in the Lung After Intranasal Vaccination With Murine Cytomegalovirus. Front Immunol 2018; 9:1861. [PMID: 30154789 PMCID: PMC6102355 DOI: 10.3389/fimmu.2018.01861] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/27/2018] [Indexed: 12/30/2022] Open
Abstract
Tissue-resident memory T (TRM) cells provide first-line defense against invading pathogens encountered at barrier sites. In the lungs, TRM cells protect against respiratory infections, but wane more quickly than TRM cells in other tissues. This lack of a sustained TRM population in the lung parenchyma explains, at least in part, why infections with some pathogens, such as influenza virus and respiratory syncytial virus (RSV), recur throughout life. Intranasal (IN) vaccination with a murine cytomegalovirus (MCMV) vector expressing the M protein of RSV (MCMV-M) has been shown to elicit robust populations of CD8+ TRM cells that accumulate over time and mediate early viral clearance. To extend this finding, we compared the inflationary CD8+ T cell population elicited by MCMV-M vaccination with a conventional CD8+ T cell population elicited by an MCMV vector expressing the M2 protein of RSV (MCMV-M2). Vaccination with MCMV-M2 induced a population of M2-specific CD8+ TRM cells that waned rapidly, akin to the M2-specific CD8+ TRM cell population elicited by infection with RSV. In contrast to the natural immunodominance profile, however, coadministration of MCMV-M and MCMV-M2 did not suppress the M-specific CD8+ T cell response, suggesting that progressive expansion was driven by continuous antigen presentation, irrespective of the competitive or regulatory effects of M2-specific CD8+ T cells. Moreover, effective viral clearance mediated by M-specific CD8+ TRM cells was not affected by the coinduction of M2-specific CD8+ T cells. These data show that memory inflation is required for the maintenance of CD8+ TRM cells in the lungs after IN vaccination with MCMV.
Collapse
Affiliation(s)
- Kaitlyn M Morabito
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States.,Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, DC, United States
| | - Tracy J Ruckwardt
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Erez Bar-Haim
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States.,Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Deepika Nair
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Syed M Moin
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Alec J Redwood
- Institute for Immunology and Infectious Diseases, Murdoch University, Perth, WA, Australia
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom.,Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Barney S Graham
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
47
|
Abstract
Memory inflation, as a term, has been used for 15 years now to describe the longitudinal development of stable, expanded CD8+ T memory pools with a distinct phenotype and functional profile which emerge in specific infection and vaccine settings. These settings have in common the persistence of antigen, especially cytomegalovirus infection but also more recently adenoviral vector vaccination. However, in contrast to chronic infections which lead to "exhaustion" the repeated antigen encounters experienced by CD8+ T cells lead to development of a robust T-cell population structure which maintains functionality and size. In this review, I will discuss how the ideas around this form of memory have evolved over time and some new models which can help explain how these populations are induced and sustained. These models are relevant to immunity against persistent viruses, to novel vaccine strategies and to concepts about aging.
Collapse
Affiliation(s)
- Paul Klenerman
- Peter Medawar Building for Pathogen Research and Translational Gastroenterology UnitUniversity of OxfordOxfordUK
| |
Collapse
|
48
|
Baumann NS, Torti N, Welten SPM, Barnstorf I, Borsa M, Pallmer K, Oduro JD, Cicin-Sain L, Ikuta K, Ludewig B, Oxenius A. Tissue maintenance of CMV-specific inflationary memory T cells by IL-15. PLoS Pathog 2018; 14:e1006993. [PMID: 29652930 PMCID: PMC5919076 DOI: 10.1371/journal.ppat.1006993] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 04/25/2018] [Accepted: 03/27/2018] [Indexed: 12/16/2022] Open
Abstract
Cytomegalovirus (CMV) infection induces an atypical CD8 T cell response, termed inflationary, that is characterised by accumulation and maintenance of high numbers of effector memory like cells in circulation and peripheral tissues—a feature being successfully harnessed for vaccine purposes. Although stability of this population depends on recurrent antigen encounter, the requirements for prolonged survival in peripheral tissues remain unknown. Here, we reveal that murine CMV-specific inflationary CD8 T cells are maintained in an antigen-independent manner and have a half-life of 12 weeks in the lung tissue. This half-life is drastically longer than the one of phenotypically comparable inflationary effector cells. IL-15 alone, and none of other common γ-cytokines, was crucial for survival of inflationary cells in peripheral organs. IL-15, mainly produced by non-hematopoietic cells in lung tissue and being trans-presented, promoted inflationary T cell survival by increasing expression of Bcl-2. These results indicate that inflationary CD8 T cells are not just simply effector-like cells, rather they share properties of both effector and memory CD8 T cells and they appear to be long-lived cells compared to the effector cells from acute virus infections. A majority of the human population is infected with cytomegalovirus (CMV), which results in lifelong persistence due to viral latency. CMV induces remarkably strong and sustained effector memory-like CD8 T cell responses in circulation and peripheral tissues, also referred to as memory CD8 T cell "inflation". In tissues, these effector memory-like cells contribute to immunosurveillance and early control of CMV reactivation events. Due to the high numbers and effector-like functional properties of inflationary CD8 T cells in peripheral tissues, CMV-based vectors are gaining substantial interest in the context of T cell based vaccines that protect peripheral tissues against infections or tumors. Here, we investigated how the stable peripheral pool of inflationary CD8 T cells is maintained and show that inflationary CD8 T cells are long-lived T cells and have a markedly prolonged half-life compared to effector CD8 T cells. In peripheral organs such as lung, IL-15 cytokine is pivotal in promoting maintenance of inflationary cells by inducing expression of the anti-apoptotic molecule Bcl-2. We show that IL-15 is mainly expressed by non-hematopoietic cells in lung tissue and that IL-15 is trans-presented to the inflationary CD8 T cells in vivo. Thus, CMV-driven inflationary CD8 T cell responses represent a unique T cell subset in peripheral tissues that is regulated differently compared to TRM CD8 T cells emerging after vaccination or acute infections.
Collapse
Affiliation(s)
- Nicolas S. Baumann
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Nicole Torti
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Suzanne P. M. Welten
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Isabel Barnstorf
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Mariana Borsa
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Katharina Pallmer
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Jennifer D. Oduro
- Department of Vaccinology and applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Luka Cicin-Sain
- Department of Vaccinology and applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Koichi Ikuta
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Annette Oxenius
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
- * E-mail:
| |
Collapse
|
49
|
Delpoux A, Michelini RH, Verma S, Lai CY, Omilusik KD, Utzschneider DT, Redwood AJ, Goldrath AW, Benedict CA, Hedrick SM. Continuous activity of Foxo1 is required to prevent anergy and maintain the memory state of CD8 + T cells. J Exp Med 2017; 215:575-594. [PMID: 29282254 PMCID: PMC5789410 DOI: 10.1084/jem.20170697] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 10/18/2017] [Accepted: 12/06/2017] [Indexed: 12/11/2022] Open
Abstract
Delpoux et al. show, in a model of latent infection, how FOXO1 is required to prevent apoptosis, the acquisition of an anergy phenotype, and to be constantly expressed for maintaining the differentiation state of CD8+ T cells. Upon infection with an intracellular pathogen, cytotoxic CD8+ T cells develop diverse differentiation states characterized by function, localization, longevity, and the capacity for self-renewal. The program of differentiation is determined, in part, by FOXO1, a transcription factor known to integrate extrinsic input in order to specify survival, DNA repair, self-renewal, and proliferation. At issue is whether the state of T cell differentiation is specified by initial conditions of activation or is actively maintained. To study the spectrum of T cell differentiation, we have analyzed an infection with mouse cytomegalovirus, a persistent-latent virus that elicits different cytotoxic T cell responses characterized as acute resolving or inflationary. Our results show that FOXO1 is continuously required for all the phenotypic characteristics of memory-effector T cells such that with acute inactivation of the gene encoding FOXO1, T cells revert to a short-lived effector phenotype, exhibit reduced viability, and manifest characteristics of anergy.
Collapse
Affiliation(s)
- Arnaud Delpoux
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla CA.,Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA
| | - Rodrigo Hess Michelini
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla CA.,Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA
| | - Shilpi Verma
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Chen-Yen Lai
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla CA.,Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA
| | - Kyla D Omilusik
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla CA
| | - Daniel T Utzschneider
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla CA.,Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA
| | - Alec J Redwood
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
| | - Ananda W Goldrath
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla CA
| | - Chris A Benedict
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Stephen M Hedrick
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla CA .,Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA
| |
Collapse
|
50
|
Cicin-Sain L, Arens R. Exhaustion and Inflation at Antipodes of T Cell Responses to Chronic Virus Infection. Trends Microbiol 2017; 26:498-509. [PMID: 29249600 DOI: 10.1016/j.tim.2017.11.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 11/22/2017] [Accepted: 11/30/2017] [Indexed: 12/21/2022]
Abstract
Viruses that have coevolved with their host establish chronic infections that are well tolerated by the host. Other viruses, that are partly adapted to their host, may induce chronic infections where persistent replication and viral antigen expression occur. The former induce highly functional and resilient CD8T cell responses called memory inflation. The latter induce dysfunctional and exhausted responses. The reasons compelling T cell responses towards inflationary or exhausted responses are only partly understood. In this review we compare the two conditions and describe mechanistic similarities and differences. We also provide a list of potential reasons why exhaustion or inflation occur in different virus infections. We propose that T cell-mediated transcriptional repression of viral gene expression provides a critical feature of inflation that allows peaceful virus and host coexistence. The virus is controlled, but its genome is not eradicated. If this mechanism is not available, as in the case of RNA viruses, the virus and the host are compelled to an arms race. If virus proliferation and spread proceed uncontrolled for too long, T cells are forced to strike a balance between viral control and tissue destruction, losing antiviral potency and facilitating virus persistence.
Collapse
Affiliation(s)
- Luka Cicin-Sain
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany; Institute for Virology, Medical School Hannover, Hannover, Germany; German Center for Infection Research (DZIF), Partner site Hannover/Braunschweig, Germany.
| | - Ramon Arens
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|